Functional Genomics Identify a Regulatory Risk Variation Rs4420550 in the 16P11.2 Schizophrenia- Associated Locus

Total Page:16

File Type:pdf, Size:1020Kb

Functional Genomics Identify a Regulatory Risk Variation Rs4420550 in the 16P11.2 Schizophrenia- Associated Locus Biological Psychiatry Archival Report Functional Genomics Identify a Regulatory Risk Variation rs4420550 in the 16p11.2 Schizophrenia- Associated Locus Hong Chang, Xin Cai, Hui-Juan Li, Wei-Peng Liu, Li-Juan Zhao, Chu-Yi Zhang, Jun-Yang Wang, Jie-Wei Liu, Xiao-Lei Ma, Lu Wang, Yong-Gang Yao, Xiong-Jian Luo, Ming Li, and Xiao Xiao ABSTRACT BACKGROUND: Genome-wide association studies (GWASs) have reported hundreds of genomic loci associated with schizophrenia, yet identifying the functional risk variations is a key step in elucidating the underlying mechanisms. METHODS: We applied multiple bioinformatics and molecular approaches, including expression quantitative trait loci analyses, epigenome signature identification, luciferase reporter assay, chromatin conformation capture, homology- directed genome editing by CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/Cas9), RNA sequencing, and ATAC-Seq (assay for transposase-accessible chromatin using sequencing). RESULTS: We found that the schizophrenia GWAS risk variations at 16p11.2 were significantly associated with messenger RNA levels of multiple genes in human brain, and one of the leading expression quantitative trait loci genes, MAPK3, is located w200 kb away from these risk variations in the genome. Further analyses based on the epigenome marks in human brain and cell lines suggested that a noncoding single nucleotide polymorphism, rs4420550 (p=2.36 3 1029 in schizophrenia GWAS), was within a DNA enhancer region, which was validated via in vitro luciferase reporter assays. The chromatin conformation capture experiment showed that the rs4420550 region physically interacted with the MAPK3 promoter and TAOK2 promoter. Precise CRISPR/Cas9 editing of a single base pair in cells followed by RNA sequencing further confirmed the regulatory effects of rs4420550 on the transcription of 16p11.2 genes, and ATAC-Seq demonstrated that rs4420550 affected chromatin accessibility at the 16p11.2 region. The rs4420550-[A/A] cells showed significantly higher proliferation rates compared with rs4420550-[G/G] cells. CONCLUSIONS: These results together suggest that rs4420550 is a functional risk variation, and this study illustrates an example of comprehensive functional characterization of schizophrenia GWAS risk loci. https://doi.org/10.1016/j.biopsych.2020.09.016 Accumulating evidence suggests that schizophrenia is a highly MAPK3). Other studies also reported regulatory effects of heritable polygenic disorder, and genome-wide surveys have 16p11.2 CNV genes on dendritic spine development and reported more than 100 risk loci containing common single synaptic function (10–12), which are believed to contribute to nucleotide polymorphisms (SNPs) (1,2) or rare copy number the pathogenesis of schizophrenia (13–18). variations (CNVs) (3–6). Among them, the chromosome Common variations in the proxy 16p11.2 CNV region also 16p11.2 region has been consistently highlighted in genetic showed genome-wide significant associations with schizo- studies of schizophrenia. For example, rare microduplications phrenia (1,2,19). However, risk SNPs primarily locate in non- at the recurrent w600-kb 16p11.2 BP4-BP5 locus are signifi- coding regions with undetermined physiological impact. It is cantly associated with schizophrenia (4,5,7). This 16p11.2 CNV presumed that such noncoding DNA variations should either region encompasses more than 25 genes. A previous study be in linkage with unknown causal mutations or exert regula- examined the effect of each individual gene on zebrafish head tory effects themselves (20–27), e.g., they might affect sizes and identified KCTD13 as the major driver, which messenger RNA (mRNA) expression through modulating participated in head-size regulation by interacting with MAPK3 physical interaction between transcription factors and and MVP (8). Besides, Blizinsky et al. (9) observed increased sequence elements (e.g., DNA enhancers) (28–34). In the pre- dendritic arborization in cortical pyramidal neurons isolated sent study, we performed brain expression quantitative trait from mice carrying the heterozygous 16p11.2 duplication, loci (eQTLs) analyses and found that schizophrenia risk SNPs which was rescued through pharmacological targeting of at 16p11.2 were significantly associated with mRNA expres- ERK-1 (extracellular signal-regulated kinase 1) (encoded by sion of multiple genes. Combinatory analyses including the 246 ª 2020 Society of Biological Psychiatry. Biological Psychiatry February 1, 2021; 89:246–255 www.sobp.org/journal ISSN: 0006-3223 Biological Molecular Mechanisms Underlying Genetic Risk at 16p11.2 Psychiatry epigenome signature identification, luciferase reporter assay, was normalized to that of Renilla luciferase to control for chromatin conformation capture (3C), homology-directed transfection efficiency variations. All assays were performed in genome editing by CRISPR/Cas9 (clustered regularly inter- triplicate in at least three independent experiments. Two-tailed spaced short palindromic repeats/Cas9), RNA sequencing t test was performed for statistical analyses. (RNA-Seq), and ATAC-Seq (assay for transposase-accessible The 3C assay was performed following previous studies chromatin using sequencing) identified a functional noncoding (48,49) to detect physical interactions between genomic re- SNP rs4420550 residing in an enhancer region at 16p11.2. gions. A total of 1 3 107 HEK293T cells in 10 mL of Dulbecco’s Modified Eagle Medium culture medium were fixed using 1% formaldehyde at room temperature for 10 minutes, and the METHODS AND MATERIALS fixed cells were lysed in 5 mL lysis buffer for 15 minutes at 4C The study protocol was approved by the Institutional Review with gentle pipetting. The mixture was centrifuged, and the Board of Kunming Institute of Zoology, Chinese Academy of nuclei pellet was then suspended in 500 mL of 1.23 restriction Sciences. Detailed methods and materials are described in the enzyme buffer. Following this, the pellets were digested with Methods and Materials in Supplement 1. The primer se- 400 U restriction enzyme EcoRI (New England Biolabs, Ips- quences in real-time quantitative polymerase chain reaction wich, MA), and T4 DNA ligase (100 U; Promega) was added to (RT-qPCR) analysis are shown in Table S1 in Supplement 2, the diluted chromatin and incubated for 4 hours at 16C. the primers used in the 3C assay are listed in Table S2 in Proteinase K (500 mg) was then added and incubated at 65C Supplement 2, the single guide RNA oligo sequences targeting overnight to reverse crosslinks, and RNA was removed genes are shown in Table S3 in Supplement 2, and the through RNase A (300 mg) treatment for 60 minutes at 37C. nucleotide sequences of the double-stranded oligonucleotides TaqMan RT-qPCR (Thermo Fisher Scientific) was performed to in electrophoretic mobility shift assay are shown in Table S4 in determine the interaction frequencies between target sites of Supplement 2. interest, and the PCR products were purified using a QIAGEN The eQTL analyses were performed using datasets of the quick gel purification kit for Sanger sequencing of each dorsolateral prefrontal cortex (DLPFC) tissues from BrainSeq chimeric DNA (QIAGEN, Hilden, Germany). To normalize primer Phase 1 (n=412) (35),BrainxQTL(n=494) (36), Common- efficiency, control PCR templates were generated by digestion Mind Consortium (n=467) (37), Genotype-Tissue Expression and random ligation of bacterial artificial chromosomes (GTEx) Project (n=175) (38), and PsychENCODE Consortium covering the 16p11.2 region. (n=1387) (39). We applied the summary data–based Homology-directed repair–mediated genome editing of Mendelian randomization (SMR) analysis (40,41) to prioritize rs4420550 by CRISPR/Cas9 was conducted in HEK293T cells risk genes, which integrated the schizophrenia genome-wide following a previous study with modifications (50). The proto- association studies (GWASs) (40,675 cases and 64,643 spacer sequence of CRISPR/Cas9 targeting rs4420550 (50- controls) (2) with the aforementioned eQTL datasets to assess GATGTTGCTAGGAGCTGACC-30) was designed using Opti- the pleiotropic effects of SNPs on diagnosis and mRNA mized CRISPR Design (http://crispr.mit.edu), and annealed olig- expressions. omers were cloned into the pL-CRISPR-E.EFS.GFP plasmid. A Linkage disequilibrium (LD) between pairwise SNPs was single-stranded oligodeoxynucleotide (50-gttttttgtgaaattattttgct- calculated using Haploview v4.1 (42) based on the European caacaggacaccaccatggccgtgctgttggtgccaggCcagctcctagcaa- genotype data from 1000 Genomes Project (43). Histone catcaaggcttctggagtgagggtgcaaaccagctcccagctggcagc-30)was marks (such as H3K4me1, H3K4me3, and H3K27ac) were synthesized as a homology-directed repair template for precise queried in the available Roadmap Epigenomics projects and editing of the rs4420550 locus. The pL-CRISPR-E.EFS.GFP ENCODE (Encyclopedia of DNA Elements) datasets of brain construct and the single-stranded oligodeoxynucleotide tissues and cell lines (HEK293 [human embryonic kidney 293] were transiently cotransfected into HEK293T cells on 100-mm and SK-N-SH) (44,45). RegulomeDB (http://www.regulomedb. plates using Lipofectamine 3000. Forty-eight hours after org/)(46) and HaploReg v4.1 (https://pubs.broadinstitute.org/ transfection, cells positive for green fluorescent protein were mammals/haploreg/haploreg.php)(47) were also used to sorted with a flow cytometer. Colonies derived from single determine the SNPs overlapped with open chromatin peaks cells were then obtained from these cells, and four
Recommended publications
  • KCTD13 Is a Major Driver of Mirrored Neuroanatomical Phenotypes Associated with the 16P11.2 CNV
    KCTD13 is a Major Driver of Mirrored Neuroanatomical Phenotypes Associated with the 16p11.2 CNV The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters. Citation Golzio, Christelle, Jason Willer, Michael E. Talkowski, Edwin C. Oh, Yu Taniguchi, Sébastien Jacquemont, Alexandre Reymond, et al. 2012. KCTD13 is a major driver of mirrored neuroanatomical phenotypes associated with the 16p11.2 CNV. Nature 485(7398): 363-367. Published Version doi:10.1038/nature11091 Accessed February 19, 2015 11:53:26 AM EST Citable Link http://nrs.harvard.edu/urn-3:HUL.InstRepos:10579139 Terms of Use This article was downloaded from Harvard University's DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http://nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA (Article begins on next page) NIH Public Access Author Manuscript Nature. Author manuscript; available in PMC 2012 November 16. Published in final edited form as: Nature. ; 485(7398): 363–367. doi:10.1038/nature11091. KCTD13 is a major driver of mirrored neuroanatomical phenotypes associated with the 16p11.2 CNV $watermark-text $watermark-text $watermark-text Christelle Golzio1, Jason Willer1, Michael E Talkowski2,3, Edwin C Oh1, Yu Taniguchi5, Sébastien Jacquemont4, Alexandre Reymond6, Mei Sun2, Akira Sawa5, James F Gusella2,3, Atsushi Kamiya5, Jacques S Beckmann4,7, and Nicholas Katsanis1,8 1Center for Human Disease Modeling and Dept of Cell biology,
    [Show full text]
  • Transcriptome Analyses of Rhesus Monkey Pre-Implantation Embryos Reveal A
    Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press Transcriptome analyses of rhesus monkey pre-implantation embryos reveal a reduced capacity for DNA double strand break (DSB) repair in primate oocytes and early embryos Xinyi Wang 1,3,4,5*, Denghui Liu 2,4*, Dajian He 1,3,4,5, Shengbao Suo 2,4, Xian Xia 2,4, Xiechao He1,3,6, Jing-Dong J. Han2#, Ping Zheng1,3,6# Running title: reduced DNA DSB repair in monkey early embryos Affiliations: 1 State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China 2 Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China 3 Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China 4 University of Chinese Academy of Sciences, Beijing, China 5 Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China 6 Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China * Xinyi Wang and Denghui Liu contributed equally to this work 1 Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press # Correspondence: Jing-Dong J. Han, Email: [email protected]; Ping Zheng, Email: [email protected] Key words: rhesus monkey, pre-implantation embryo, DNA damage 2 Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press ABSTRACT Pre-implantation embryogenesis encompasses several critical events including genome reprogramming, zygotic genome activation (ZGA) and cell fate commitment.
    [Show full text]
  • Capra Hircus)
    ARTICLES OPEN Sequencing and automated whole-genome optical mapping of the genome of a domestic goat (Capra hircus) Yang Dong1,2,12, Min Xie3,12, Yu Jiang1,5,12, Nianqing Xiao10,12, Xiaoyong Du4,12, Wenguang Zhang1,6,12, Gwenola Tosser-Klopp7, Jinhuan Wang1, Shuang Yang3, Jie Liang3, Wenbin Chen3, Jing Chen3, Peng Zeng3, Yong Hou3, Chao Bian3, Shengkai Pan3, Yuxiang Li3, Xin Liu3, Wenliang Wang3, Bertrand Servin7, Brian Sayre11, Bin Zhu10, Deacon Sweeney10, Rich Moore10, Wenhui Nie1, Yongyi Shen1,2, Ruoping Zhao1, Guojie Zhang3, Jinquan Li6, Thomas Faraut7, James Womack9, Yaping Zhang1, James Kijas5, Noelle Cockett8, Xun Xu1–3, Shuhong Zhao4, Jun Wang3 & Wen Wang1 We report the ~2.66-Gb genome sequence of a female Yunnan black goat. The sequence was obtained by combining short-read sequencing data and optical mapping data from a high-throughput whole-genome mapping instrument. The whole-genome mapping data facilitated the assembly of super-scaffolds >5× longer by the N50 metric than scaffolds augmented by fosmid end sequencing (scaffold N50 = 3.06 Mb, super-scaffold N50 = 16.3 Mb). Super-scaffolds are anchored on chromosomes based on conserved synteny with cattle, and the assembly is well supported by two radiation hybrid maps of chromosome 1. We annotate 22,175 protein-coding genes, most of which were recovered in the RNA-seq data of ten tissues. Comparative transcriptomic analysis of the primary and secondary follicles of a cashmere goat reveal 51 genes that are differentially expressed between the two types of hair follicles. This study, whose results will facilitate goat genomics, shows that whole-genome mapping technology can be used for the de novo assembly of large genomes.
    [Show full text]
  • Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights Into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle
    animals Article Integrating Single-Step GWAS and Bipartite Networks Reconstruction Provides Novel Insights into Yearling Weight and Carcass Traits in Hanwoo Beef Cattle Masoumeh Naserkheil 1 , Abolfazl Bahrami 1 , Deukhwan Lee 2,* and Hossein Mehrban 3 1 Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; [email protected] (M.N.); [email protected] (A.B.) 2 Department of Animal Life and Environment Sciences, Hankyong National University, Jungang-ro 327, Anseong-si, Gyeonggi-do 17579, Korea 3 Department of Animal Science, Shahrekord University, Shahrekord 88186-34141, Iran; [email protected] * Correspondence: [email protected]; Tel.: +82-31-670-5091 Received: 25 August 2020; Accepted: 6 October 2020; Published: 9 October 2020 Simple Summary: Hanwoo is an indigenous cattle breed in Korea and popular for meat production owing to its rapid growth and high-quality meat. Its yearling weight and carcass traits (backfat thickness, carcass weight, eye muscle area, and marbling score) are economically important for the selection of young and proven bulls. In recent decades, the advent of high throughput genotyping technologies has made it possible to perform genome-wide association studies (GWAS) for the detection of genomic regions associated with traits of economic interest in different species. In this study, we conducted a weighted single-step genome-wide association study which combines all genotypes, phenotypes and pedigree data in one step (ssGBLUP). It allows for the use of all SNPs simultaneously along with all phenotypes from genotyped and ungenotyped animals. Our results revealed 33 relevant genomic regions related to the traits of interest.
    [Show full text]
  • Downloaded from ENSEMBL (Version 72), and the Reference Genome Sequence of the Goat Was Download from (CGD) (Dong Et Al
    bioRxiv preprint doi: https://doi.org/10.1101/743955; this version posted August 22, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Convergent genomic signatures of high altitude adaptation among domestic mammals Dong-Dong Wu 1, 5, 10, *, ‡, Cui-Ping Yang 3,5 ‡, Ming-Shan Wang 1, 5, ‡, Kun-Zhe Dong 4, ‡, Da-Wei Yan 2, ‡, Zi-Qian Hao 11, ‡, Song-Qing Fan7, Shu-Zhou Chu7, Qiu-Shuo Shen 3,5, Li-Ping Jiang 3,5, Yan Li 1, 5, Lin Zeng 1, 5, He-Qun Liu 1, 5, Hai-Bing Xie 1, 5, Yun-Fei Ma 1, 5, Xiao-Yan Kong 2, Shu-Li Yang 2, Xin-Xing Dong 2, Ali Esmailizadeh Koshkoiyeh 8, David M Irwin 1, 9, Xiao Xiao 3, Ming Li 3, Yang Dong 1, 5, Wen Wang 1, 5, Peng Shi 1, 5, Hai-Peng Li 10, 11, Yue-Hui Ma 4, *, Xiao Gou 2, *, Yong-Bin Chen 3, 5, 10, *, Ya-Ping Zhang 1, 5, 6, 10, * 1. State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China 2. Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China; 3. Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China 4. Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China. 5. Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China 6.
    [Show full text]
  • Supp Material.Pdf
    Supplementary Information Estrogen-mediated Epigenetic Repression of Large Chromosomal Regions through DNA Looping Pei-Yin Hsu, Hang-Kai Hsu, Gregory A. C. Singer, Pearlly S. Yan, Benjamin A. T. Rodriguez, Joseph C. Liu, Yu-I Weng, Daniel E. Deatherage, Zhong Chen, Julia S. Pereira, Ricardo Lopez, Jose Russo, Qianben Wang, Coral A. Lamartiniere, Kenneth P. Nephew, and Tim H.-M. Huang S1 Method Immunofluorescence staining Approximately 2,000 mammosphere-derived epithelial cells (MDECs) cells seeded collagen I-coated coverslips were fixed with methanol/acetone for 10 min. After blocking with 2.5% bovine serum albumin (Sigma) for 1 hr, these cells were incubated with anti-ESR1 antibody (Santa Cruz) overnight at 4˚C. The corresponding secondary FITC-conjugated antibody was applied followed by DAPI staining (Molecular Probes) for the nuclei. Photographs were captured by Zeiss fluorescence microscopy (Zeiss). The percentages of ESR1 subcellular localization were calculated in ten different optical fields (~10 cells per field) by two independent researchers. References Carroll, J.S., Meyer, C.A., Song, J., Li, W., Geistlinger, T.R., Eeckhoute, J., Brodsky, A.S., Keeton, E.K., Fertuck, K.C., Hall, G.F., et al. 2006. Genome-wide analysis of estrogen receptor binding sites. Nat. Genet. 38: 1289-1297. Neve, R.M., Chin, K., Fridlyand, J., Yeh, J., Baehner, F.L., Fevr, T., Clark, L., Bayani, N., Coppe, J.P., Tong, F., et al. 2006. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10: 515-527. S2 Hsu et al. Supplementary Information A Figure S1. Integrative mapping of large genomic regions subjected to ERα-mediated epigenetic repression.
    [Show full text]
  • Mutpred2: Inferring the Molecular and Phenotypic Impact of Amino Acid Variants
    bioRxiv preprint doi: https://doi.org/10.1101/134981; this version posted May 9, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. MutPred2: inferring the molecular and phenotypic impact of amino acid variants Vikas Pejaver1, Jorge Urresti2, Jose Lugo-Martinez1, Kymberleigh A. Pagel1, Guan Ning Lin2, Hyun-Jun Nam2, Matthew Mort3, David N. Cooper3, Jonathan Sebat2, Lilia M. Iakoucheva2, Sean D. Mooney4, and Predrag Radivojac1 1Indiana University, Bloomington, Indiana, U.S.A. 2University of California San Diego, La Jolla, California, U.S.A. 3Cardiff University, Cardiff, U.K. 4University of Washington, Seattle, Washington, U.S.A. Contact: [email protected]; [email protected]; [email protected] Abstract We introduce MutPred2, a tool that improves the prioritization of pathogenic amino acid substitutions, generates molecular mechanisms potentially causative of disease, and returns interpretable pathogenicity score distributions on individual genomes. While its prioritization performance is state-of-the-art, a novel and distinguishing feature of MutPred2 is the probabilistic modeling of variant impact on specific as- pects of protein structure and function that can serve to guide experimental studies of phenotype-altering variants. We demonstrate the utility of MutPred2 in the iden- tification of the structural and functional mutational signatures relevant to Mendelian disorders and the prioritization of de novo mutations associated with complex neu- rodevelopmental disorders. We then experimentally validate the functional impact of several variants identified in patients with such disorders. We argue that mechanism- driven studies of human inherited diseases have the potential to significantly accelerate the discovery of clinically actionable variants.
    [Show full text]
  • Associated 16P11.2 Deletion in Drosophila Melanogaster
    ARTICLE DOI: 10.1038/s41467-018-04882-6 OPEN Pervasive genetic interactions modulate neurodevelopmental defects of the autism- associated 16p11.2 deletion in Drosophila melanogaster Janani Iyer1, Mayanglambam Dhruba Singh1, Matthew Jensen1,2, Payal Patel 1, Lucilla Pizzo1, Emily Huber1, Haley Koerselman3, Alexis T. Weiner 1, Paola Lepanto4, Komal Vadodaria1, Alexis Kubina1, Qingyu Wang 1,2, Abigail Talbert1, Sneha Yennawar1, Jose Badano 4, J. Robert Manak3,5, Melissa M. Rolls1, Arjun Krishnan6,7 & 1234567890():,; Santhosh Girirajan 1,2,8 As opposed to syndromic CNVs caused by single genes, extensive phenotypic heterogeneity in variably-expressive CNVs complicates disease gene discovery and functional evaluation. Here, we propose a complex interaction model for pathogenicity of the autism-associated 16p11.2 deletion, where CNV genes interact with each other in conserved pathways to modulate expression of the phenotype. Using multiple quantitative methods in Drosophila RNAi lines, we identify a range of neurodevelopmental phenotypes for knockdown of indi- vidual 16p11.2 homologs in different tissues. We test 565 pairwise knockdowns in the developing eye, and identify 24 interactions between pairs of 16p11.2 homologs and 46 interactions between 16p11.2 homologs and neurodevelopmental genes that suppress or enhance cell proliferation phenotypes compared to one-hit knockdowns. These interac- tions within cell proliferation pathways are also enriched in a human brain-specific network, providing translational relevance in humans. Our study indicates a role for pervasive genetic interactions within CNVs towards cellular and developmental phenotypes. 1 Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA. 2 Bioinformatics and Genomics Program, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
    [Show full text]
  • Long-Term Propagation of Tree Shrew Spermatogonial Stem Cells in Culture and Successful Generation of Transgenic Offspring
    Cell Research (2017) 27:241-252. © 2017 IBCB, SIBS, CAS All rights reserved 1001-0602/17 $ 32.00 ORIGINAL ARTICLE www.nature.com/cr Long-term propagation of tree shrew spermatogonial stem cells in culture and successful generation of transgenic offspring Chao-Hui Li1, 2, 3, Lan-Zhen Yan4, 5, Wen-Zan Ban1, Qiu Tu4, 3, Yong Wu4, 3, Lin Wang1, 2, Rui Bi4, Shuang Ji1, Yu-Hua Ma5, Wen-Hui Nie1, Long-Bao Lv5, Yong-Gang Yao3, 4, 5, 6, Xu-Dong Zhao4, 5, Ping Zheng1, 2, 5 1State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; 2Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; 3Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; 4Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Scienc- es, Kunming 650223, China; 5Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; 6CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China Tree shrews have a close relationship to primates and have many advantages over rodents in biomedical research. However, the lack of gene manipulation methods has hindered the wider use of this animal. Spermatogonial stem cells (SSCs) have been successfully expanded in culture to permit sophisticated gene editing in the mouse and rat. Here, we describe a culture system for the long-term expansion of tree shrew SSCs without the loss of stem cell properties.
    [Show full text]
  • Targeting the RHOA Pathway Improves Learning and Memory In
    bioRxiv preprint doi: https://doi.org/10.1101/2020.05.22.110098; this version posted May 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Targeting the RHOA pathway improves learning and memory in Kctd13 and 16p11.2 deletion mouse models. Authors Sandra MARTIN LORENZO1, Valérie NALESSO1, Claire CHEVALIER1, Marie-Christine BIRLING2, and Yann HERAULT1,2,*. Affiliations 1 Université de Strasbourg, CNRS, INSERM, Institut de Génétique Biologie Moléculaire et Cellulaire, IGBMC - UMR 7104 - Inserm U1258, 1 rue Laurent Fries, 67404 ILLKIRCH cedex 2 Université de Strasbourg, CNRS, INSERM, CELPHEDIA-PHENOMIN-ICS, Institut Clinique de la Souris, 1 rue Laurent Fries, 67404 ILLKIRCH cedex *Corresponding author: Yann HERAULT, [email protected] Keywords: Copy number variation, neurodevelopment, intellectual disability, autism spectrum disorders, KCTD13, mouse model, recognition memory. Running title: Therapeutic effects of fasudil in 16p11.2 models. 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.05.22.110098; this version posted May 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. HIGHLIGHTS - Kctd13 haploinsufficiency recapitulates most of the behaviour phenotypes found in the 16p11.2 Del/+ models - Fasudil treatment restores Kctd13 and 16p11.2 Del/+ mutant phenotypes in novel location and novel object recognition memory tests - Fasudil treatment restores the RhoA pathway in Kctd13+/- and 16p11.2 Del/+ models ABSTRACT Gene copy number variants (CNV) have an important role in the appearance of neurodevelopmental disorders.
    [Show full text]
  • SZDB: a Database for Schizophrenia Genetic Research
    Schizophrenia Bulletin vol. 43 no. 2 pp. 459–471, 2017 doi:10.1093/schbul/sbw102 Advance Access publication July 22, 2016 SZDB: A Database for Schizophrenia Genetic Research Yong Wu1,2, Yong-Gang Yao1–4, and Xiong-Jian Luo*,1,2,4 1Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China; 2Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China; 3CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China 4YGY and XJL are co-corresponding authors who jointly directed this work. *To whom correspondence should be addressed; Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; tel: +86-871-68125413, fax: +86-871-68125413, e-mail: [email protected] Schizophrenia (SZ) is a debilitating brain disorder with a Introduction complex genetic architecture. Genetic studies, especially Schizophrenia (SZ) is a severe mental disorder charac- recent genome-wide association studies (GWAS), have terized by abnormal perceptions, incoherent or illogi- identified multiple variants (loci) conferring risk to SZ. cal thoughts, and disorganized speech and behavior. It However, how to efficiently extract meaningful biological affects approximately 0.5%–1% of the world populations1 information from bulk genetic findings of SZ remains a and is one of the leading causes of disability worldwide.2–4 major challenge. There is a pressing
    [Show full text]
  • Cul3 Regulates Cytoskeleton Protein Homeostasis and Cell Migration During a Critical
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.10.902064; this version posted January 11, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Cul3 regulates cytoskeleton protein homeostasis and cell migration during a critical 2 window of brain development 3 Jasmin Morandell1, Lena A. Schwarz1, Bernadette Basilico1, Saren Tasciyan1, Armel Nicolas1, 4 Christoph Sommer1, Caroline Kreuzinger1, Christoph P. Dotter1, Lisa S. Knaus1, Zoe Dobler1, 5 Emanuele Cacci2, Johann G. Danzl1, Gaia Novarino1@ 6 7 1Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria 8 2 Department of Biology and Biotechnology “Charles Darwin”, Sapienza, University of Rome 9 @Correspondence to: [email protected] 10 11 Abstract 12 De novo loss of function mutations in the ubiquitin ligase-encoding gene Cullin3 (CUL3) lead to 13 autism spectrum disorder (ASD). Here, we used Cul3 mouse models to evaluate the 14 consequences of Cul3 mutations in vivo. Our results show that Cul3 haploinsufficient mice 15 exhibit deficits in motor coordination as well as ASD-relevant social and cognitive impairments. 16 Cul3 mutant brain displays cortical lamination abnormalities due to defective neuronal migration 17 and reduced numbers of excitatory and inhibitory neurons. In line with the observed abnormal 18 columnar organization, Cul3 haploinsufficiency is associated with decreased spontaneous 19 excitatory and inhibitory activity in the cortex. At the molecular level, employing a quantitative 20 proteomic approach, we show that Cul3 regulates cytoskeletal and adhesion protein abundance 21 in mouse embryos.
    [Show full text]